Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Clin Chim Acta ; 508: 221-227, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32422129

RESUMEN

Vascular wall calcification (VC) is highly prevalent in patients with chronic kidney disease (CKD). In CKD, VC is more frequent and severe than in the general population and it is associated with increased cardiovascular mortality and morbidity. In the last years, laboratory and clinical evidence have drawn the attention to the relationship between bone disease and VC in CKD patients, leading to the concept of a bone-vessel or bone-vascular axis. It means that disorders of bone volume and bone turnover may influence the risk of VC and ultimately the high risk of cardiovascular mortality. In fact, a higher burden of VC has been associated to low bone volume and low bone turnover in hemodialysis (HD) patients with renal osteodystrophy characterized by histomorphometric evaluation of bone biopsies. The molecular mechanisms underlying the regulation of bone cells and vascular cells in CKD are poorly understood. In this review, we discuss relevant evidence linking bone disorders and VC in CKD and also rising molecular players involved in this bone-vascular axis. Indeed, accumulating data is available for two proposed systems: receptor activator for nuclear factor kB (RANK)/ RANK ligand (RANKL)/osteoprotegerin (OPG) system and inhibitors of Wnt signaling - mainly sclerostin. Although they are promising biochemical markers linking bone formation and bone reabsorption with VC, there is a long way to go as long evidence from laboratory studies is often divergent to the clinical data as will be discussed. Future prospective studies are needed in order to evaluate the role of these biochemical players as useful clinical markers for VC, bone volume and perhaps bone turnover.


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica , Insuficiencia Renal Crónica , Calcificación Vascular , Huesos , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/diagnóstico , Humanos , Laboratorios , Estudios Prospectivos
2.
Kidney Int ; 95(5): 1064-1078, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30878213

RESUMEN

Calcimimetics decrease parathyroid hormone (PTH) secretion in patients with secondary hyperparathyroidism. The decrease in PTH should cause a reduction in bone turnover; however, the direct effect of calcimimetics on bone cells, which express the calcium-sensing receptor (CaSR), has not been defined. In this study, we evaluated the direct bone effects of CaSR activation by a calcimimetic (AMG 641) in vitro and in vivo. To create a PTH "clamp," total parathyroidectomy was performed in rats with and without uremia induced by 5/6 nephrectomy, followed by a continuous subcutaneous infusion of PTH. Animals were then treated with either the calcimimetic or vehicle. Calcimimetic administration increased osteoblast number and osteoid volume in normal rats under a PTH clamp. In uremic rats, the elevated PTH concentration led to reduced bone volume and increased bone turnover, and calcimimetic administration decreased plasma PTH. In uremic rats exposed to PTH at 6-fold the usual replacement dose, calcimimetic administration increased osteoblast number, osteoid surface, and bone formation. A 9-fold higher dose of PTH caused an increase in bone turnover that was not altered by the administration of calcimimetic. In an osteosarcoma cell line, the calcimimetic induced Erk1/2 phosphorylation and the expression of osteoblast genes. The addition of a calcilytic resulted in the opposite effect. Moreover, the calcimimetic promoted the osteogenic differentiation and mineralization of human bone marrow mesenchymal stem cells in vitro. Thus, calcimimetic administration has a direct anabolic effect on bone that counteracts the decrease in PTH levels.


Asunto(s)
Compuestos de Bifenilo/administración & dosificación , Remodelación Ósea/efectos de los fármacos , Calcimiméticos/administración & dosificación , Hiperparatiroidismo Secundario/tratamiento farmacológico , Fallo Renal Crónico/complicaciones , Fenetilaminas/administración & dosificación , Animales , Modelos Animales de Enfermedad , Humanos , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/etiología , Masculino , Osteoblastos/efectos de los fármacos , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/sangre , Hormona Paratiroidea/metabolismo , Ratas , Ratas Wistar , Receptores Sensibles al Calcio/metabolismo
3.
Clin Kidney J ; 11(1): 80-88, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29423207

RESUMEN

Secondary hyperparathyroidism (SHPT) is associated with increased bone turnover, risk of fractures, vascular calcifications, and cardiovascular and all-cause mortality. The classical treatment for SHPT includes active vitamin D compounds and phosphate binders. However, achieving the optimal laboratory targets is often difficult because vitamin D sterols suppress parathyroid hormone (PTH) secretion, while also promoting calcium and phosphate intestinal absorption. Calcimimetics increase the sensitivity of the calcium-sensing receptor, so that even with lower levels of extracellular calcium a signal can still exist, leading to a decrease of the set-point for systemic calcium homeostasis. This enables a decrease in plasma PTH levels and, consequently, of calcium levels. Cinacalcet was the first calcimimetic to be approved for clinical use. More than 10 years since its approval, cinacalcet has been demonstrated to effectively reduce PTH and improve biochemical control of mineral and bone disorders in chronic kidney patients. Three randomized controlled trials have analysed the effects of treatment with cinacalcet on hard clinical outcomes such as vascular calcification, bone histology and cardiovascular mortality and morbidity. However, a final conclusion on the effect of cinacalcet on hard outcomes remains elusive. Etelcalcetide is a new second-generation calcimimetic with a pharmacokinetic profile that allows thrice-weekly dosing at the time of haemodialysis. It was recently approved in Europe, and is regarded as a second opportunity to improve outcomes by optimizing treatment for SHPT. In this review, we summarize the impact of cinacalcet with regard to biochemical and clinical outcomes. We also discuss the possible implications of the new calcimimetic etelcalcetide in the quest to improve outcomes.

4.
PLoS One ; 12(7): e0179868, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28686736

RESUMEN

BACKGROUND AND OBJECTIVES: In dialysis patients, there is an increasing evidence that altered bone metabolism is associated with cardiovascular calcifications. The main objective of this study was to analyse, in hemodialysis patients, the relationships between bone turnover, mineralization and volume, evaluated in bone biopsies, with a plain X-ray vascular calcification score. DESIGN, SETTING, PARTICIPANTS AND MEASUREMENTS: In a cross-sectional study, bone biopsies and evaluation of vascular calcifications were performed in fifty hemodialysis patients. Cancellous bone volume, mineralized bone volume, osteoid volume, activation frequency, bone formation rate/bone surface, osteoid thickness and mineralization lag time were determined by histomorphometry. Vascular calcifications were assessed by the simple vascular calcification score (SVCS) in plain X-Ray of pelvis and hands and, for comparison, by the Agatston score in Multi-Slice Computed Tomography (MSCT). RESULTS: SVCS≥3 was present in 20 patients (40%). Low and high bone turnover were present in 54% and 38% of patients, respectively. Low bone volume was present in 20% of patients. In multivariable analysis, higher age (p = 0.015) and longer hemodialysis duration (p = 0.017) were associated with SVCS≥3. Contrary to cancellous bone volume, the addition to this model of mineralized bone volume (OR = 0.863; 95%CI: 0.766, 0.971; p = 0.015), improved the performance of the model. For each increase of 1% in mineralized bone volume there was a 13.7% decrease in the odds of having SVCS≥3 (p = 0.015). An Agatston score>400 was observed in 80% of the patients with a SVCS≥3 versus 4% of patients with a SVCS<3, (p<0.001). CONCLUSION: Higher mineralized bone volume was associated with a lower plain X-ray vascular calcification. This study corroborates the hypothesis of the existence of a link between bone and vessel and reinforces the clinical utility of this simple and inexpensive vascular calcification score in dialysis patients.


Asunto(s)
Calcificación Fisiológica , Diálisis Renal/efectos adversos , Tomografía Computarizada de Emisión , Calcificación Vascular/diagnóstico por imagen , Adulto , Anciano , Biopsia , Desarrollo Óseo/fisiología , Femenino , Mano/diagnóstico por imagen , Mano/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Pelvis/diagnóstico por imagen , Pelvis/fisiopatología , Calcificación Vascular/diagnóstico , Calcificación Vascular/fisiopatología
5.
Clin Kidney J ; 8(5): 567-71, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26413282

RESUMEN

Calcific uraemic arteriolopathy (CUA) is a rare disease and continues to be a clinical challenge. The typical course of CUA is characterized by painful skin discolouration and induration evolving to necrotic ulcerations. Medial calcification of cutaneous arterioles and extensive extracellular matrix remodelling are the hallmarks of CUA. The epidemiology and risk factors associated with this disease are still not fully understood. Moreover, CUA treatment strategies vary significantly among centres and expert recommendations are heterogeneous. Registries may provide important insights and information to increase our knowledge about epidemiology and clinical aspects of CUA and may help to optimize its therapeutic management. In 2006, we established an internet-based registry in Germany (www.calciphylaxie.de) to allow online notification of patients with established or suspected CUA. The registry includes a comprehensive database with questions covering >70 parameters and items regarding patient-related and laboratory data, clinical background and presentation as well as therapeutic strategies. The next phase will be to allow international patient registration via www.calciphylaxis.net as part of the multinational EuCalNet (European Calciphylaxis Network) initiative, which is supported by the ERA-EDTA scientific working group 'CKD-MBD'. Based on the valuable experience with the previous German CUA registry, EuCalNet will be a useful tool to collect data on the rare disease CUA and may become a basis for prospective controlled trials in the near future.

6.
J Nephrol ; 28(5): 531-40, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25835730

RESUMEN

Calciphylaxis, or calcific uremic arteriolopathy, is a vascular ossification-calcification disease involving cutaneous or visceral arterioles, with ischemic damage of the surrounding tissues, usually in the setting of chronic kidney disease. Pathogenesis is still unclear and probably comprises the participation of vascular smooth muscle cells, endothelial cells and macrophages surrounded by a uremic and/or pro-calcifying environment. According to the original concept of calcific uremic arteriolopathy coined by Hans Selye, risk factors may be divided into sensitizers and challengers and their knowledge is useful in clinical practice to pre-emptively identify both uremic and non-uremic 'at risk' patients and guide treatment. Systemic calcific uremic arteriolopathy is a rarity. Cutaneous calcific uremic arteriolopathy is more frequent and clinically presents as a first phase of cutaneous hardening and erythema, followed by a second phase of ulcerations and scars; these two phases are probably associated with the initial development of arteriolar lesion and tissue ischemic damage, respectively. Clinical history, physical examination, laboratory analysis, histology and imaging are the main tools to exclude important differential diagnoses and obtain a definitive diagnosis. Treatment is generally unrewarding and consists of rigorous control of comorbid conditions, anti-oxidant, anti-inflammatory and antithrombotic strategies, avoidance of iatrogeny and wound and pain management. Prognosis remains poor in terms of morbidity and mortality. Efforts should be made towards a greater awareness of calcific uremic arteriolopathy, development of better therapies and improvement of clinical outcomes.


Asunto(s)
Calcifilaxia , Diagnóstico por Imagen/métodos , Manejo de la Enfermedad , Fallo Renal Crónico/complicaciones , Calcifilaxia/diagnóstico , Calcifilaxia/etiología , Calcifilaxia/terapia , Progresión de la Enfermedad , Humanos , Factores de Riesgo
7.
Nefrología (Madr.) ; 33(6): 835-844, nov.-dic. 2013. ilus
Artículo en Inglés | IBECS | ID: ibc-121412

RESUMEN

Fibroblast Growth Factor 23 (FGF-23) is a bone-derived hormone involved in the regulation of phosphate homeostasis. FGF-23 levels are extremely elevated in Chronic Kidney Disease (CKD) and there is evidence supporting the role of this hormone in the pathogenesis of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Furthermore, recent data associates FGF-23 with the pathogenesis of systemic complications of CKD-MBD. The increasing evidence that the consequences of abnormal mineral metabolism are not restricted to bone disease changed the approach to the pathophysiology and treatment of disturbed bone and mineral metabolism in CKD patients. FGF-23 has been proposed to be the initial adaptive response in early CKD to protect the organism from the adverse effects of phosphate retention. Increased levels of FGF-23 observed in CKD patients are associated with cardiovascular mortality risk and was shown to mediate direct, "off-target" toxicity to the heart. This report aims to review the relevant aspects of the physiology of FGF-23 in bone biology and mineral homeostasis and the role of FGF-23 in the pathophysiology of CKD-BMD and its clinical implications (AU)


El factor de crecimiento fibroblástico 23 (FGF-23) es una hormona derivada del hueso que participa en la regulación de la homeostasis del fósforo. Los niveles de FGF-23 se encuentran extremamente elevados en la enfermedad renal crónica y existe evidencia del papel de esta hormona en la patogénesis de los trastornos óseos y minerales en esta situación. Más aún, datos recientes implican al FGF-23 en la patogénesis de otras complicaciones sistémicas asociadas a las alteraciones óseo-minerales de la enfermedad renal crónica. La evidencia creciente de que las alteraciones del metabolismo mineral no se limitan a la enfermedad ósea ha acentuado el interés por la patofisiología y el tratamiento de las alteraciones del metabolismo mineral en la enfermedad renal crónica. Se ha propuesto que el aumento de FGF-23 es la respuesta inicial en los estadios precoces de la enfermedad renal crónica a la necesidad de proteger al organismo de los efectos adversos de la retención de fósforo. Estos aumentos de FGF-23 se asocian al riesgo creciente de mortalidad cardiovascular en los enfermos renales crónicos y son mediadores directos de toxicidad cardíaca. En esta revisión procuramos presentar aspectos relevantes de la fisiología del FGF-23 en la biología ósea y en la homeostasis mineral, así como en la fisiopatología de la enfermedad renal crónica y sus implicaciones clínicas (AU)


Asunto(s)
Humanos , Factores de Crecimiento de Fibroblastos/análisis , Insuficiencia Renal Crónica/fisiopatología , Calcificación Fisiológica/fisiología , Desmineralización Ósea Patológica/fisiopatología
8.
Nefrologia ; 33(6): 835-44, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24158124

RESUMEN

Fibroblast Growth Factor 23 (FGF-23) is a bone-derived hormone involved in the regulation of phosphate homeostasis. FGF-23 levels are extremely elevated in Chronic Kidney Disease (CKD) and there is evidence supporting the role of this hormone in the pathogenesis of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Furthermore, recent data associates FGF-23 with the pathogenesis of systemic complications of CKD-MBD. The increasing evidence that the consequences of abnormal mineral metabolism are not restricted to bone disease changed the approach to the pathophysiology and treatment of disturbed bone and mineral metabolism in CKD patients. FGF-23 has been proposed to be the initial adaptive response in early CKD to protect the organism from the adverse effects of phosphate retention. Increased levels of FGF-23 observed in CKD patients are associated with cardiovascular mortality risk and was shown to mediate direct, "off-target" toxicity to the heart. This report aims to review the relevant aspects of the physiology of FGF-23 in bone biology and mineral homeostasis and the role of FGF-23 in the pathophysiology of CKD-BMD and its clinical implications.


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/fisiopatología , Factores de Crecimiento de Fibroblastos/fisiología , Insuficiencia Renal Crónica/fisiopatología , Animales , Huesos/metabolismo , Calcio/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/etiología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Progresión de la Enfermedad , Retroalimentación Fisiológica , Factor-23 de Crecimiento de Fibroblastos , Homeostasis , Humanos , Hierro/fisiología , Ratones , Modelos Biológicos , Osteocitos/metabolismo , Hormona Paratiroidea/fisiología , Fósforo/metabolismo , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Resultado del Tratamiento , Vitamina D/análogos & derivados , Vitamina D/biosíntesis , Vitamina D/fisiología
9.
BMC Nephrol ; 13: 76, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863242

RESUMEN

BACKGROUND: Elevated serum phosphorus (P) levels have been linked to increased morbidity and mortality in dialysis patients with secondary hyperparathyroidism (SHPT) but may be difficult to control if parathyroid hormone (PTH) is persistently elevated. We conducted a post hoc analysis of data from an earlier interventional study (OPTIMA) to explore the relationship between PTH control and serum P. METHODS: The OPTIMA study randomized dialysis patients with intact PTH (iPTH) 300-799 pg/mL to receive conventional care alone (vitamin D and/or phosphate binders [PB]; n=184) or a cinacalcet-based regimen (n=368). For patients randomized to conventional care, investigators were allowed flexibility in using a non-cinacalcet regimen (with no specific criteria for vitamin D analogue dosage) to attain KDOQI™ targets for iPTH, P, Ca and Ca x P. For those assigned to the cinacalcet-based regimen, dosages of cinacalcet, vitamin D sterols, and PB were optimized over the first 16 weeks of the study, using a predefined treatment algorithm. The present analysis examined achievement of serum P targets (≤ 4.5 and ≤ 5.5 mg/dL) in relation to achievement of iPTH ≤ 300 pg/mL during the efficacy assessment phase (EAP; weeks 17-23). RESULTS: Patients who achieved iPTH ≤ 300 pg/mL (or a reduction of ≥ 30% from baseline) were more likely to achieve serum P targets than those who did not, regardless of treatment group. Of those who did achieve iPTH ≤ 300 pg/mL, 43% achieved P ≤ 4.5 mg/dL and 70% achieved P ≤ 5.5 mg/dL, versus 21% and 46% of those who did not achieve iPTH ≤ 300 pg/mL. Doses of PB tended to be higher in patients not achieving serum P targets. Patients receiving cinacalcet were more likely to achieve iPTH ≤ 300 pg/mL than those receiving conventional care (73% vs 23% of patients). Logistic regression analysis identified lower baseline P, no PB use at baseline and cinacalcet treatment to be predictors of achieving P ≤ 4.5 mg/dL during EAP in patients above this threshold at baseline. CONCLUSIONS: This post hoc analysis found that control of serum P in dialysis patients was better when serum PTH levels were lowered effectively, regardless of treatment received. TRIAL REGISTRATION: Clinicaltrials.gov identifier NCT00110890.


Asunto(s)
Diálisis/estadística & datos numéricos , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/epidemiología , Hormona Paratiroidea/sangre , Fósforo/sangre , Insuficiencia Renal/sangre , Insuficiencia Renal/rehabilitación , Adulto , Anciano , Causalidad , Comorbilidad , Europa (Continente)/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Insuficiencia Renal/epidemiología , Factores de Riesgo
10.
Nephron Clin Pract ; 120(2): c108-19, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22555359

RESUMEN

Phosphate-binder therapy for hyperphosphataemia is key to the treatment of patients with chronic kidney disease (CKD)-mineral and bone disorder (MBD). Calcium-free phosphate binders are increasingly favoured since calcium-based agents potentially cause harmful calcium overload and vascular calcification that confound the benefits of reducing serum phosphorus. Several calcium-free phosphate binders are available, including the non-absorbed agent sevelamer and the absorbed agents, e.g. lanthanum and magnesium salts. Randomised controlled studies consistently show that sevelamer and lanthanum carbonate offer equivalent lowering of serum phosphorus and often effectively achieve phosphorus targets versus calcium salts, with sevelamer having a positive effect on bone disease, vascular calcification, and patient-level outcomes in dialysis patients in several trials. There is also evidence that lanthanum carbonate can improve bone health, but data are limited to its effects to vascular calcification or patient-level outcomes. Magnesium salts have also been shown to reduce serum phosphorus levels, but clear evidence is lacking on bone, vascular, or clinical outcomes. It also remains to be established whether long-term systemic accumulation of lanthanum and magnesium, in tissues including bone, has clinically relevant toxic effects. This review summarises the evidence of efficacy and safety for newer calcium-free phosphate binders in CKD-MBD management.


Asunto(s)
Quelantes/química , Quelantes/uso terapéutico , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/tratamiento farmacológico , Medicina Basada en la Evidencia , Hiperfosfatemia/tratamiento farmacológico , Fallo Renal Crónico/tratamiento farmacológico , Fosfatos/química , Calcio/química , Quelantes/efectos adversos , Humanos , Hiperfosfatemia/complicaciones , Fallo Renal Crónico/complicaciones
11.
Curr Opin Nephrol Hypertens ; 18(4): 303-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19424062

RESUMEN

PURPOSE OF REVIEW: Adynamic bone disease has recently been associated with increased risk of vascular calcification. This review focuses on the emerging data in adynamic bone disease, its clinical consequences and therapeutic implications. RECENT FINDINGS: There is a lack of good biochemical markers of parathyroid status, bone formation and reabsorption to allow a secure diagnosis of bone disease. Recent data have suggested a possible link between bone activity and vascular calcification. Cardiovascular calcification is an independent predictor of mortality. Adynamic bone is associated with a very low capacity of bone to incorporate calcium in the bone compartment and inability to handle an extra calcium load. A positive association between vascular calcifications and low bone turnover has been suggested. Calcium-containing phosphate binders, active vitamin D therapy and high calcium dialysate may enhance vascular calcifications in the presence of adynamic bone disease. SUMMARY: There is recent evidence suggesting a negative impact of calcium load in the progression of vascular calcification in dialysis patients with chronic kidney disease stage 5 with adynamic bone disease. The current therapeutic approach to these patients should focus on reduction of calcium and vitamin D load to restore parathyroid activity.


Asunto(s)
Enfermedades Óseas/etiología , Enfermedades Óseas/terapia , Animales , Enfermedades Óseas/fisiopatología , Remodelación Ósea , Calcinosis/etiología , Calcio/metabolismo , Enfermedad Crónica , Enfermedad de la Arteria Coronaria/etiología , Humanos , Enfermedades Renales/complicaciones , Hormona Paratiroidea/sangre , Diálisis Renal/efectos adversos
12.
Nephron Clin Pract ; 112(1): c41-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19365139

RESUMEN

BACKGROUND/AIMS: The calcimimetic cinacalcet (Mimpara/Sensipar) simultaneously lowers parathyroid hormone (PTH), phosphorus (P) and calcium (Ca) levels in patients with secondary hyperparathyroidism. The OPTIMA study demonstrated that cinacalcet and adjusted doses of vitamin D maximized control of these parameters. This post-hoc analysis of OPTIMA data assessed the impact of reducing or increasing the dose of concomitant vitamin D on PTH, P and Ca in patients receiving cinacalcet. METHODS: Dialysis patients with mean baseline intact PTH (iPTH) 300-800 pg/ml (31.8-84.8 pM) received doses of cinacalcet titrated to achieve an iPTH of 150-300 pg/ml (15.9-31.8 pM). The dose of vitamin D could then be decreased to further reduce serum P or Ca, or increased/initiated to further decrease PTH levels if iPTH >300 pg/ml or to increase Ca if Ca <8.0 mg/dl (2.0 mM). RESULTS: Vitamin D dose was assessed for 345 patients during a 23-week period. A total of 91 and 129 patients had an increase or decrease in vitamin D dose, respectively. By study end, mean iPTH, P, and Ca were similar in both vitamin D groups, although there were differences in biochemical parameters between groups at the start of the study. There were statistically significant reductions from baseline to study end in iPTH and Ca in both groups (p < 0.001). Although P was significantly reduced by week 23 in the group in which vitamin D dose was decreased (p = 0.007), the reduction in P was less and did not achieve significance in the group in which vitamin D dose was increased (p = 0.71). CONCLUSIONS: After initiating cinacalcet, the dose of vitamin D can be adjusted to maximize reductions in PTH, P and Ca; however, vitamin D-induced decreases in PTH need to be balanced with the diminished response in P and Ca.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Naftalenos/uso terapéutico , Vitamina D/administración & dosificación , Adulto , Calcio/sangre , Cinacalcet , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hipercalcemia/tratamiento farmacológico , Hipercalcemia/etiología , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/etiología , Hiperfosfatemia/tratamiento farmacológico , Hiperfosfatemia/etiología , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Masculino , Hormona Paratiroidea/sangre , Fósforo/sangre , Diálisis Renal , Vitamina D/farmacología , Vitamina D/uso terapéutico
13.
Clin J Am Soc Nephrol ; 4(2): 450-5, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19158372

RESUMEN

BACKGROUND AND OBJECTIVES: There is increasing evidence that altered bone metabolism is associated with cardiovascular calcifications in patients with stage 5 chronic kidney disease on hemodialysis (HD). This study was conducted to evaluate the association between bone volume, turnover, and coronary calcifications in HD patients. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: In a cross-sectional study, bone biopsies and multislice computed tomography were performed in 38 HD patients. Bone volume/total volume, activation frequency, and bone formation rate/bone surface were determined by histomorphometry and coronary calcifications were quantified by Agatston scores. RESULTS: Prevalence of low bone turnover was 50% and of low bone volume was 16%. Among the studied traditional cardiovascular risk factors, only age was found to be associated with coronary calcifications. Lower bone volume was a significant risk factor for coronary calcifications during early years of HD, whereas this effect was not observed in patients with dialysis duration >6 yr. Histomorphometric parameters of bone turnover were not associated with coronary calcifications. CONCLUSIONS: Low bone volume is associated with increased coronary calcifications in patients on HD.


Asunto(s)
Remodelación Ósea , Calcinosis/etiología , Enfermedad de la Arteria Coronaria/etiología , Ilion/patología , Enfermedades Renales/terapia , Diálisis Renal/efectos adversos , Adulto , Factores de Edad , Anciano , Biopsia , Calcinosis/patología , Calcinosis/fisiopatología , Enfermedad Crónica , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/fisiopatología , Estudios Transversales , Femenino , Humanos , Ilion/diagnóstico por imagen , Ilion/fisiopatología , Enfermedades Renales/complicaciones , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Tamaño de los Órganos , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Tomografía Computarizada por Rayos X , Adulto Joven
14.
Kidney Int ; 74(12): 1505-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19034298

RESUMEN

In dialysis patients, there is an association between vascular calcifications and mortality. Hyperphosphatemia and calcium overload are associated with development of vascular calcifications, especially in the presence of low bone turnover. Different plain X-ray methods are now available to evaluate vascular calcifications in dialysis patients. The presence of vascular calcifications is an alert sign for increased cardiovascular risk, and this information is important for choosing the most suitable treatment for dialysis patients.


Asunto(s)
Calcinosis/diagnóstico por imagen , Enfermedades Cardiovasculares/etiología , Enfermedades Renales/complicaciones , Calcinosis/etiología , Enfermedades Cardiovasculares/diagnóstico por imagen , Humanos , Hiperfosfatemia/etiología , Radiografía , Diálisis Renal , Riesgo
15.
Kidney Int Suppl ; (111): S38-43, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19034324

RESUMEN

In chronic kidney disease patients, bone and mineral abnormalities have a major impact on morbidity and mortality. Hyperphosphatemia has been associated with increased mortality and with the development of cardiovascular calcification, an independent predictor of mortality. Vascular calcifications have been associated with low bone turnover, low bone volume and lower activation frequency. In dialysis patients, the treatment of hyperphospathemia with calcium based compounds, when compared with sevelamer, is associated with more frequent episodes of hypercalcemia, suppression of intact parathyroid hormone and with progression of coronary calcifications. In the presence of adynamic bone disease, calcium load has a significantly higher impact on aortic calcifications and stiffening. A randomized, prospective, open label study, evaluated patients with bone biopsies at the beginning and after 1 year treatment period with sevelamer hydrochloride or calcium carbonate. Sevelamer treatment resulted in no statistically significant changes in bone turnover or mineralization compared with calcium carbonate, but bone formation rate increased and trabecular architecture improved only with sevelamer. In incident dialysis patients, treatment with sevelamer has been associated with better survival, while in prevalent patients a clear benefit could only be demonstrated in older patients and in patients treated for more than 2 years.In conclusion, the treatment of hyperphosphatemia with sevelamer hydrochloride, a non-calcium and non-metal containing phosphate binder, is associated with a beneficial effect on vascular calcification progression, bone disease and most likely with a survival benefit in some hemodialysis patients populations.


Asunto(s)
Hiperfosfatemia/tratamiento farmacológico , Poliaminas/uso terapéutico , Diálisis Renal , Enfermedades Óseas/prevención & control , Calcinosis/prevención & control , Humanos , Hiperfosfatemia/complicaciones , Hiperfosfatemia/etiología , Enfermedades Renales/complicaciones , Enfermedades Renales/mortalidad , Sevelamer , Análisis de Supervivencia , Enfermedades Vasculares/prevención & control
17.
Nephrol Dial Transplant ; 23(9): 2889-94, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18398016

RESUMEN

BACKGROUND: Secondary hyperparathyroidism (SHPT) is a common complication of chronic kidney disease. Increased parathyroid hormone (PTH) synthesis and secretion is associated with parathyroid cell hyperplasia. The exact mechanisms involved in parathyroid gland (PTG) hyperplasia are still poorly understood. There is no available data on angiogenesis in PTG of patients with chronic kidney disease and SHPT. The aim of this study is to evaluate angiogenesis and expression of the angiogenic factors, basic fibroblastic growth factor (b-FGF) and vascular endothelial growth factor A (VEGF), in secondary PTG hyperplasia. METHODS: This study was performed on formalin-fixed paraffin-embedded archival tissues of 21 SHPT glands from haemodialysis (n = 19) and kidney transplanted (n = 2) patients submitted to surgical parathyroidectomy. For control, eight normal human parathyroid glands (NPG) encountered in surgical specimens of total thyroidectomy were used. We evaluated the immunohistochemical expression of the proliferation cell marker Ki67. Angiogenesis was evaluated by immunohistochemistry staining with anti-endoglin (CD105) antibody in 21 SPH and 5 NPG by stereological analysis. Levels of b-FGF and VEGF were determined by semi-quantitative analysis in 21 SPH and 8 NPG. RESULTS: The SHPT patients present a mean iPTH of 1314 +/- 750 pg/ml, a corrected serum calcium of 10.3 +/- 1.2 mg/dl and a serum phosphorus of 6.1 +/- 1.4 mg/dl. SHPT glands displayed a significantly higher immunoreactivity against Ki67, compared to NPG. With CD105, a significantly higher number and volume of microvessels were observed in SHPT compared to NPG. Both VEGF and b-FGF expression were increased in SHPT compared to NPG. Using the predefined subdivision into negative and positive only the b-FGF expression was significantly increased in the SHPT glands compared to NPG. CONCLUSION: These results suggest that PTGs in this group of patients with SHPT have a significantly higher number of vessels expressing CD105, which has been reported to preferentially label activated endothelial cells associated with angiogenesis. SHPT glands have a significantly increased expression of b-FGF compared to NPG. VEGF-A expression is also increased in the examined SHPT glands but could be less relevant for angiogenesis.


Asunto(s)
Hiperparatiroidismo Secundario/fisiopatología , Neovascularización Patológica/metabolismo , Glándulas Paratiroides/patología , Insuficiencia Renal Crónica/patología , Anciano , Antígenos CD/metabolismo , Endoglina , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Hiperparatiroidismo Secundario/metabolismo , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Superficie Celular/metabolismo , Insuficiencia Renal Crónica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Clin J Am Soc Nephrol ; 3(1): 36-45, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18178780

RESUMEN

BACKGROUND AND OBJECTIVES: Cinacalcet, a novel calcimimetic, targets the calcium-sensing receptor to lower parathyroid hormone (PTH), calcium, and phosphorus levels in dialysis patients with secondary hyperparathyroidism (SHPT). This study compared the efficacy of a cinacalcet-based regimen with unrestricted conventional care (vitamin D and phosphate binders) for achieving the stringent National Kidney Foundation Kidney Disease Outcomes Quality Initiative (KDOQI) targets for dialysis patients. STUDY DESIGN: In this multicenter, open-label study, hemodialysis patients with poorly controlled SHPT were randomized to receive conventional care (n = 184) or a cinacalcet-based regimen (n = 368). Doses of cinacalcet, vitamin D sterols, and phosphate binders were adjusted during a 16-wk dose-optimization phase with the use of algorithms that allowed cinacalcet to be used with adjusted doses of vitamin D. The primary end point was the proportion of patients with mean intact PTH < or =300 pg/ml during a 7-wk efficacy assessment phase. RESULTS: A higher proportion of patients receiving the cinacalcet-based regimen versus conventional care achieved the targets for PTH (71% versus 22%, respectively; P < 0.001), Ca x P (77% versus 58%, respectively; P < 0.001), calcium (76% versus 33%, respectively; P < 0.001), phosphorus (63% versus 50%, respectively; P = 0.002), and PTH and Ca x P (59% versus 16%, respectively, P < 0.001), and allowed a 22% reduction in vitamin D dosage in patients receiving vitamin D at baseline. Achievement of targets was greatest in patients with less severe disease (intact PTH range, 300 to 500 pg/ml) and the cinacalcet dose required was lower in these patients (median = 30 mg/d). CONCLUSIONS: Compared with conventional therapy, a cinacalcet-based treatment algorithm increased achievement of KDOQI treatment targets in dialysis patients in whom conventional therapy was no longer effective in controlling this disease.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Fallo Renal Crónico/complicaciones , Naftalenos/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , Calcio/sangre , Quelantes/administración & dosificación , Quelantes/efectos adversos , Cinacalcet , Femenino , Humanos , Hiperparatiroidismo Secundario/sangre , Masculino , Persona de Mediana Edad , Naftalenos/efectos adversos , Hormona Paratiroidea/sangre , Fósforo/sangre , Resultado del Tratamiento , Vitamina D/administración & dosificación , Vitamina D/efectos adversos
19.
Kidney Int Suppl ; (85): S91-6, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12753275

RESUMEN

BACKGROUND: Recognition of the role of the extracellular calcium-sensing receptor (CaR) in mineral metabolism has greatly improved our understanding of calcium homeostasis. The activation of this receptor by small changes in the extracellular ionized calcium (ec(Ca2+)) regulates PTH, calcitonin secretion, urinary calcium excretion, and, ultimately, bone turnover. METHODS: The cloning of the CaR and the discovery of mutations that make the receptor less or more sensitive to calcium have allowed a better understanding of several hereditary disorders characterized by either hyperparathyroidism or hypoparathyroidism. The CaR, able to amplify the sensitivity of the CaR to Ca++ and suppress PTH levels with a resulting decrease in blood Ca++, became an ideal target for the development of compounds, the calcimimetics. Experience with the calcimimetic R-568 in patients with primary and secondary hyperparathyroidism and parathyroid carcinoma are summarized. RESULTS: The first clinical studies with the first-generation calcimimetic agents have demonstrated their efficacy in lowering plasma intact PTH concentration in uremic patients with secondary hyperparathyroidism. However, the low bioavailability of these first calcimimetics predicts a difficult clinical utilization. The second-generation calcimimetic, AMG 073, having a better pharmacokinetic profile, appears to be effective and safe for the treatment of secondary hyperparathyroidism, suppressing PTH levels while simultaneously reducing serum phosphorus levels and the calcium x phosphorus product. CONCLUSION: The advantage of controlling PTH secretion without the complications related to hypercalcemia, hyperphosphatemia, and increased calcium x phosphorus product is very promising.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Calcio/fisiología , Hiperparatiroidismo Secundario/tratamiento farmacológico , Receptores Sensibles al Calcio/fisiología , Compuestos de Anilina/farmacocinética , Animales , Ensayos Clínicos como Asunto , Humanos , Fallo Renal Crónico/complicaciones , Hormona Paratiroidea/metabolismo , Fenetilaminas , Propilaminas , Receptores Sensibles al Calcio/efectos de los fármacos
20.
Kidney Int Suppl ; (80): 149-54, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11982829

RESUMEN

Recognition of the role of the extracellular calcium sensing receptor (CaR) in mineral metabolism has greatly improved our understanding of calcium homeostasis. The biology of the low affinity, G-protein-coupled CaR and the effects of its activation in various tissues are reviewed. Physiological roles include regulation of parathyroid hormone (PTH) secretion by small changes in ionized calcium (Ca++), and control of urinary calcium excretion with small changes in blood Ca++. The CaR also affects the renal handling of sodium, magnesium, and water. Mutations affecting the CaR that make it either less or more sensitive to Ca++ cause various clinical disorders. Disorders, such as primary and secondary hyperparathyroidism, may exhibit acquired abnormalities of the CaR. Calcimimetic drugs, which amplify the sensitivity of the CaR to Ca++, can suppress PTH levels with a resultant fall in blood Ca++. Experiences with R-568 in patients with secondary and primary hyperparathyroidism and parathyroid carcinoma are summarized. In humans with hyperparathyroidism, these agents produce a dose-dependent fall in PTH and blood Ca++, with larger doses causing more sustained effects. The second generation calcimimetic, AMG 073, with a better pharmacokinetic profile appears to be an effective and safe treatment for secondary hyperparathyroidism, producing suppression of PTH levels with a simultaneous reduction in serum phosphorus levels and the calcium X phosphorus product. The advantage of controlling PTH secretion without the complications related to hypercalcemia, hyperphosphatemia, and increased calcium X phosphorus product is very promising. Treatment trials have been relatively short-term except for one patient treated with R-568 for more than 600 days for parathyroid carcinoma; nonetheless the drug had no major side effects and appeared to be safe. Further long-term controlled studies are underway to further confirm the effectiveness and safety of these compounds.


Asunto(s)
Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/fisiología , Proteínas de Unión al Calcio , Humanos , Hiperparatiroidismo Secundario/tratamiento farmacológico , Riñón/fisiología , Receptores Sensibles al Calcio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...